throbber
GASTROENTEROLOGY 2000;118:735–748
`
`Molecular Mechanism of Interferon Alfa–Mediated Growth
`Inhibition in Human Neuroendocrine Tumor Cells
`
`KATHARINA M. DETJEN,* MARTINA WELZEL,* KATRIN FARWIG,* FELIX H. BREMBECK,‡
`ASTRID KAISER,‡ ERNST–OTTO RIECKEN,‡ BERTRAM WIEDENMANN,* and STEFAN ROSEWICZ*
`*Medizinische Klinik mit Schwerpunkt Hepatologie und Gastroenterologie, Universita¨tsklinikum Charite´, Campus Virchow Klinikum, Humboldt
`Universita¨t zu Berlin; and ‡Innere Medizin I, Gastroenterologie und Infektiologie, Universita¨tsklinikum Benjamin Franklin, Freie Universita¨t Berlin,
`Berlin, Germany
`
`Background & Aims: Although human neuroendocrine
`tumors respond to interferon (IFN)-a treatment in vivo,
`the underlying mechanisms of growth inhibition are
`poorly understood. To characterize the antiproliferative
`effects at a molecular level, we explored the growth-
`regulatory action of IFN-a in the human neuroendo-
`crine tumor cell lines BON and QGP1. Methods: IFN-a
`receptor expression and signal transduction were exam-
`ined by reverse-transcription polymerase chain reac-
`tion, immunoblotting, subcellular fractionation, and
`transactivation assays. Growth regulation was evalu-
`ated by cell numbers, soft agar assays, and cell cycle
`analysis using flow cytometry. Expression and activity
`of cell cycle–regulatory molecules were determined by
`immunoblotting and histone H1–kinase assays. Results:
`Both cell lines expressed IFN-a receptor mRNA tran-
`scripts. Ligand binding initiated phosphorylation of Jak
`kinases and Stat transcription factors, resulting in Stat
`activation, nuclear translocation, and transcription from
`an ISRE-reporter construct. Prolonged IFN-a treatment
`dose-dependently inhibited both anchorage-depen-
`dent and -independent growth. Cell cycle analysis of
`IFN-a–treated, unsynchronized cultures revealed an
`increased S-phase population, which was further sub-
`IFN-a–
`stantiated in G1 synchronized QGP1 cells.
`treated cells entered S phase in parallel to control
`cultures, but their progress into G2/M phase was
`delayed. Both cellular cyclin B levels and CDC 2 activity
`were substantially reduced. The extent and time course
`of this reduction corresponded to the observed S-
`phase accumulation. Conclusions: IFN-a directly inhib-
`its growth of human neuroendocrine tumor cells by
`specifically delaying progression through S phase and
`into G2/M. These cell cycle changes are associated with
`inhibition of cyclin B expression, resulting in reduced
`CDC2 activity.
`
`Neuroendocrine (NE) gastroenteropancreatic (GEP)
`
`tumors constitute a biologically heterogeneous group
`of rare, predominantly slow-progressing but mostly
`malignant neoplasms.1 They frequently synthesize and
`
`secrete bioactive molecules, such as regulatory peptides,
`kinins, and serotonin, which are responsible for the
`tumor-associated hypersecretion syndrome.2 Because more
`than 80% of malignant GEP tumors present liver
`metastases at the time of diagnosis, systemic therapy is
`usually required.1
`Current therapeutic strategies aim to control both
`hypersecretion-related symptoms and tumor growth.3–5
`Somatostatin and its analogues are effective in suppress-
`ing the hypersecretion syndrome, but they are frequently
`insufficient to control tumor progression.6 Therefore, multi-
`modal therapeutic approaches are evaluated that encom-
`pass surgical debulking, chemoembolization, chemother-
`apy, and biotherapy.3,5,7 The typically low proliferative
`fraction of NE tumors limits the efficacy of conventional
`chemotherapeutic approaches, so that conclusive prolon-
`gation of survival remains to be demonstrated.8,9 In view
`of the potential side effects of chemotherapeutic regimes,
`biotherapeutic approaches using long-term treatment
`with interferon (IFN)-a alone or in combination with soma-
`tostatin analogues have widely replaced conventional
`chemotherapeutic strategies over the past decade.3–5,7,10
`Patients with advanced GEP NE tumor disease have
`received IFN-a in the context of clinical trials; a
`significant fraction of tumors responded to IFN-a treat-
`ment, as determined by biochemical markers such as
`5-hydroxyindolacetic acid excretion or chromogranin A
`serum levels.5,7,11 IFN-a–induced reduction of such
`biochemical markers was reflected by a marked relief of
`hypersecretion-associated symptoms in functionally ac-
`tive tumors. Several of these studies also reported an
`
`Abbreviations used in this paper: DMEM, Dulbecco’s modified
`Eagle medium; DTT, dithiothreitol; FCS, fetal calf serum; GED,
`gastroenteropancreatic; IFN, interferon; ISRE, interferon-stimulated
`response element; PKR, RNA-dependent protein kinase; PMSF,
`phenylmethylsulfonyl fluoride; SDS-PAGE, sodium dodecyl sulfate–
`polyacrylamide gel electrophoresis.
`r 2000 by the American Gastroenterological Association
`0016-5085/00/$10.00
`doi:10.1053/gg.2000.5968
`
`Roxane Labs., Inc.
`Exhibit 1039
`Page 001
`
`

`
`736 DETJEN ET AL.
`
`GASTROENTEROLOGY Vol. 118, No. 4
`
`IFN-a–mediated inhibition of tumor progression result-
`ing in stable disease or, in some cases, partial remission
`with reduction of tumor burden.11,12 It is of particular
`clinical relevance that inclusion of IFN-a in the therapeu-
`tic regimen enabled suppression of disease progression in
`a significant fraction of tumors resistant to monotherapy
`with somatostatin analogues.5 Nonetheless, a significant
`fraction of endocrine GEP tumors fail to respond to the
`growth-inhibitory effects of IFN-a. The clinical benefit
`of the specific antiproliferative actions of IFN-a has
`therefore remained controversial.
`Multiple mechanisms have been proposed to contrib-
`ute to overall clinical outcome of IFN-a treatment,
`including direct antiproliferative actions of IFN-a on the
`tumor cells and indirect mechanisms, such as inhibition
`of angiogenesis, induction of tumor mesenchyme, and
`immunomodulation via up-regulation of major histocom-
`patibility class I antigens (reviewed by Grander et al.13).
`Resistance of
`individual NE tumors to the growth-
`regulatory effects of IFN-a may result from defective
`signal transduction or tumor specific phenotypic alter-
`ations in growth-regulatory pathways. Because the respon-
`siveness of the secretory pathway is frequently main-
`tained in functionally active GEP tumors that are
`refractory to the growth-inhibitory actions of IFN-a,10–12
`the latter mechanism seems to be of major biological
`relevance in endocrine GEP tumors. Consequently, iden-
`tification of the specific growth-relevant cellular targets
`of IFN-a and understanding of their function are needed
`to maximally exploit the potential of IFN-a in the
`treatment of NE tumor disease.
`Much progress has been made in elucidating the signal
`transduction pathways activated by type I IFNs.14,15
`Ligand binding to specific receptors at the plasma
`membrane results in dimerization of receptor subunits
`and thereby initiates the rapid autophosphorylation of
`receptor-associated Janus tyrosine kinases ( JAKs) Jak1
`and Jak2. The activated JAKs
`in turn tyrosine-
`phosphorylate and activate latent cytosolic members of
`the Stat transcription factor family, named for their dual
`function as signal transducers and activators of transcrip-
`tion. Activated Stat1 and Stat2 transcription factors then
`associate with a 48-kilodalton protein into multimeric
`complexes termed IFN-stimulated gene factor 3 (ISGF3)
`that translocate into the nucleus where they induce
`changes in gene expression, which ultimately result in
`the biological effects of IFN-a treatment.
`In contrast to these well-established and conserved
`signal transduction events, the molecular basis of the
`antiproliferative effects of IFN-a appears to be highly cell
`type specific and has not yet been delineated in GEP NE
`tumor cells. The most comprehensive analysis of IFN-a–
`
`induced growth regulation used Daudi Burkitt lym-
`phoma cells and identified multiple signaling pathways
`that apparently function in parallel to block G1–S
`progression. Two major independent and complementary
`effector pathways have emerged from these studies. The
`first is accumulation of the hypophosphorylated, growth-
`restrictive form of the retinoblastoma-associated tumor-
`suppressor protein Rb.16–21 This functional activation of
`Rb is attributed to down-regulation of G1 cyclins and
`CDC25 phosphatases in conjunction with induction of
`cyclin-dependent kinase inhibitors (CKIs) and total Rb
`content. The second pathway is induction of the double-
`stranded RNA–activated protein kinase (p68 PKR)
`initiating down-regulation of cellular c-myc levels.22,23
`However, alternative mechanisms may prevail in NE
`tumors that have been reported to exhibit functional
`inactivation of Rb24 and overexpression of c-myc.25 To
`specifically evaluate the direct cell cycle–regulatory ef-
`fects of IFN-a on GEP NE tumor cells, we analyzed
`IFN-a–dependent signal transduction and growth regu-
`lation in an in vitro model of human GEP NE tumor
`disease.
`Materials and Methods
`Materials
`QGP1 cells were kindly provided by K. Mo¨lling
`(Zu¨ rich, Switzerland); BON cells were a generous gift from
`C. M. Townsend (Galveston, TX). Dulbecco’s modified Eagle
`medium (DMEM), RPMI 1640 medium, and phosphate-
`buffered saline (PBS) were supplied by GIBCO BRL (Berlin,
`Germany). Fetal calf serum (FCS), trypsin/EDTA, penicillin,
`and streptomycin were from Biochrom (Berlin, Germany). The
`antibodies were purchased from the following manufacturers:
`Santa Cruz Biochemicals (Santa Cruz, CA; CDK2, Jak1, Tyk2,
`Stat1, Stat2); Transduction Laboratories
`(Lexington, KY;
`p27kip1); Pharmingen (San Diego, CA; cyclins E, A, and B);
`Calbiochem–Novabiochem GmbH (Bad Soden, Germany;
`p21cip1); and Dianova GmbH (Hamburg, Germany; all second-
`ary antibodies). [g-32P]Adenosine triphosphate (ATP) and
`enhanced chemiluminescence Western blotting detection re-
`agents were obtained from Amersham (Braunschweig, Ger-
`many), and calf histone H1, deoxyribonuclease, and ATP from
`Boehringer Mannheim (Mannheim, Germany). Polymerase
`chain reaction (PCR) reagents were obtained from Promega
`(Heidelberg, Germany), and Western blot supplies from
`Bio-Rad Laboratories GmbH (Mu¨ nchen, Germany). IFN-a2b
`(Roferon) was kindly provided by Hofmann-LaRoche (Basel,
`Switzerland). Protein A–Sepharose beads and all other reagents
`were purchased from Sigma Chemical Co. (Deisenhofen, Ger-
`many).
`
`Cell Lines and Tissue Culture
`The human NE pancreatic tumor cell lines QGP1 and
`BON were grown as subconfluent monolayer cultures in RPMI
`
`Roxane Labs., Inc.
`Exhibit 1039
`Page 002
`
`

`
`April 2000
`
`IFN–a IN NEUROENDOCRINE TUMOR CELLS 737
`
`1640 and DMEM, respectively, supplemented with 10%
`(vol/vol) FCS, 100 U/mL penicillin, and 100 µg/mL streptomy-
`cin, and kept in 95% air and 5% CO2 at 37°C. Experiments
`were routinely performed in the log phase of growth after cells
`had been plated for 16–24 hours. For experiments on synchro-
`nized cultures, QGP1 cells were serum starved for 36 hours,
`which resulted in G1-phase accumulation of approximately
`70% of the cells, and were then restimulated by addition of
`10% FCS.
`
`Cell Growth Assays
`Effects of IFN-a on proliferation of NE tumor cells
`were evaluated by determination of cell numbers. Cells were
`plated at 50,000 cells/well (BON) or 25,000 cells/well (QGP1)
`in 12-well tissue culture plates, and cells were manually
`counted using a hemacytometer at indicated time points.
`Viability of cells was confirmed by trypan blue exclusion and
`was routinely .95%. To determine effects of IFN-a on
`anchorage-independent growth of NE tumor cells, colony
`formation in agar suspension was evaluated. Briefly, 3 3 104
`cells were resuspended in 300 µL of culture medium and added
`to a mixture of 2.7 mL Hyclone FCS, 0.8 mL Iscove’s modified
`DMEM, 3.6 mL of 2.1% (wt/vol) methylcellulose in Iscove’s,
`1.6 mL agar solution (10 mL 3% agar and 20 mL DMEM), and
`0.06 mL b-mercaptoethanol to obtain an agar suspension.
`Aliquots (1 mL, 3000 cells) of this suspension were then
`dispensed in 35-mm dishes containing the indicated concentra-
`tions of IFN-a or vehicle. Colony formation was assessed under
`an inverted microscope by manual counting after a 10-day
`incubation period. A threshold of 20 cells was arbitrarily set to
`score cell accumulations as colonies.
`
`Flow-Cytometric Analysis
`For cell cycle analysis, approximately 106 cells were
`harvested by gentle trypsinization (0.25%), carefully resus-
`pended in PBS, and fixed in ethanol (70%) for 30 minutes at
`–20°C. After brief centrifugation, cells were washed once with
`PBS and incubated for 30 minutes at room temperature in PBS
`containing 100 µg/mL ribonuclease A, 0.1 % Triton X-100, 1
`µmol/L EDTA, and 1.5 µg/mL propidium iodide. Cell cycle
`analysis was carried out on a FACScan using Modfit software
`(Becton Dickinson, Heidelberg, Germany).
`
`Western Blotting and Subcellular
`Fractionation
`For analysis of cell cycle proteins, subconfluent cells
`were treated as indicated, rinsed twice with ice-cold PBS
`containing 1 mmol/L sodium orthovanadate (Na3VO4), and
`extracted in ice-cold lysis buffer (20 mmol/L Tris [pH 7.8], 150
`mmol/L NaCl, 2 mmol/L EDTA, 50 mmol/L b-glycerophos-
`phate, 0.5% Nonidet P-40, 1% glycerine, 10 mmol/L NaF, 1
`mmol/L sodium orthovanadate, 1 mmol/L dithiothreitol [DTT],
`2 µmol/L phenylmethylsulfonyl fluoride [PMSF], 10 µg/mL
`aprotinin, and 2 µmol/L leupeptin). Extracts were boiled in
`Laemmli’s sample buffer, and aliquots were separated by
`sodium dodecyl sulfate–polyacrylamide gel electrophoresis
`
`(SDS-PAGE), electroblotted to polyvinyl difluoride mem-
`branes (New England Nuclear, Ko¨ln, Germany), and blocked
`in PBS/0.1% Tween (PBST) containing 5% nonfat dry milk
`(cyclins, E, A, B, p21cip1, p27kip1, Rb, p16, Jak1, Tyk2, Stat1,
`Stat2) or 5% bovine serum albumin (CDK2, CDC2) for 2 hours
`at room temperature. Incubation with primary antibodies was
`carried out overnight at 4°C and was followed by 3 washes in
`PBST at room temperature. After incubation with appropriate
`horseradish peroxidase–conjugated secondary antibodies, mem-
`branes were washed 3 times for 15 minutes in PBST, and bands
`were visualized by enhanced chemiluminescence following the
`manufacturer’s recommendations. Results were quantitated by
`laser densitometry using the Scanpak 2 software (Biometra,
`Go¨ttingen, Germany).
`For Stat translocation studies, subcellular fractions of QGP
`cells were prepared before immunoblotting according to the
`procedure described by Simboli-Campbell.26 After hypotone
`lysis (1 mmol/L NaHCO3, 5 mmol/L MgCl2, 0.1 mmol/L
`PMSF, and 20 µg/mL leupeptin), extracts were adjusted to 50
`mmol/L Tris-HCl (pH 7.5) plus 50 mmol/L EGTA and nuclei
`were isolated by brief centrifugation at 500g. Nuclei were
`further purified by saccharose gradient centrifugation (45%
`[wt/vol] saccharose, 30 minutes, 1660g) and then lysed in
`buffer A containing 50 mmol/L Tris-HCl
`(pH 7.5), 0.5
`mmol/L EGTA, 0.5 mmol/L EDTA, 1% b-mercaptoethanol, 1
`mmol/L PMSF, 20 µg/mL leupeptin, and 1% SDS by brief
`sonication. Aliquots of the nuclear and the cytosolic fractions
`were then processed as described above.
`
`Immunoprecipitation and Histone H1
`–Kinase Assays
`To determine tyrosine phosphorylation of Jak kinases
`and Stat proteins, subconfluent cells were treated as indicated,
`then washed twice in ice-cold PBS and lysed by brief sonication
`in IP buffer (20 mmol/L Tris [pH 7.8], 150 mmol/L NaCl, 2
`mmol/L EDTA, 0.5% Nonidet P-40, 10 mmol/L NaF, 10
`mmol/L sodium orthovanadate, 2 mmol/L PMSF, and 10
`µg/mL each of leupeptin, pepstatin, and aprotinin). The lysates
`were precleared for 1 hour by incubation with protein A–Sepha-
`rose beads. Immune complexes were collected on protein
`A–Sepharose beads that had been coated with saturating
`amounts of primary antibody. Beads were subsequently washed
`4 times with ice-cold IP buffer, boiled in Laemmli’s sample
`buffer, separated by SDS-PAGE, and transferred to polyvinyl
`difluoride membranes. Nonspecific binding was blocked with
`10 mmol/L Tris-HCl (pH 7.6), 150 mmol/L NaCl, and 0.05%
`Tween 20 containing bovine serum albumin (1%) for 2 hours at
`room temperature. The membrane was then incubated with
`a-phosphotyrosine antibody (1 µg/mL) for 2 hours at room
`temperature and further processed as described for Western
`blots. To control that equivalent amounts of immunocomplexes
`were analyzed, membranes were reprobed with the antibodies
`used for immunoprecipitation. For repeated use, membranes
`were stripped by incubating the membrane in a solution of
`62.5 mmol/L Tris-HCl (pH 6.8), 2% SDS, and 100 mmol/L
`2-mercaptoethanol for 30 minutes at 55°C followed by a
`
`Roxane Labs., Inc.
`Exhibit 1039
`Page 003
`
`

`
`738 DETJEN ET AL.
`
`GASTROENTEROLOGY Vol. 118, No. 4
`
`washing in PBST for 2 hours, changing the buffer every 30
`minutes.
`To examine the composition and activity of cdk complexes,
`immunoprecipitations were performed. For immunoprecipita-
`tion of CDK2 or CDC2, cells were lysed by mild sonication in
`ice-cold ELB buffer (50 mmol/L HEPES [pH 7.5], 250 mmol/L
`NaCl, 5 mmol/L EDTA, 0.1% Nonidet P-40, 1 mmol/L DTT,
`1 mmol/L NaF, 0.1 mmol/L Na3VO4, 2 µg/mL aprotinin, 5
`µg/mL leupeptin, and 0.1 mmol/L PMSF)
`followed by a
`30-minute incubation on ice with occasional vortexing. After
`brief centrifugation (10 minutes, 4°C, 13,000g), lysates (500
`µg/sample) were precleared for 1 hour by incubation with
`protein A–Sepharose beads. Immuncomplexes were then pre-
`cipitated by addition of protein A–Sepharose beads precoated
`with saturating amounts of CDK2 or CDC2 antibody (10 µg).
`Samples were incubated with gentle agitation at 4°C for 4
`hours. Immuncomplexes were subsequently washed 4 times in
`ice-cold ELB buffer and twice in 50 mmol/L HEPES (pH 7.5)
`containing 1 mmol/L DTT. The kinase reaction was then
`started by addition of 30 µL kinase buffer containing 50
`mmol/L HEPES (pH 7.5), 1 mmol/L DTT, 10 mmol/L MgCl2,
`1 µg calf histone H1 per sample, 50 µmol/L ATP, and 5 µCi
`[g-32P]ATP per sample. The kinase reactions were allowed to
`proceed for 5 minutes (CDK2) and 30 minutes (CDC2) and
`were then terminated by boiling the samples in Laemmli’s
`buffer. The whole reaction was subjected to 10% SDS-PAGE,
`and kinase activities were determined by autoradiography of
`the dried gels.
`
`Transactivation Studies
`To determine transactivation after IFN-a stimulation
`in BON NE tumor cells, transient transfection assays with an
`IFN-stimulated response element (ISRE)-luciferase reporter
`gene construct were performed. The ISRE-luc construct,
`containing residues 2206/286 of the human 28,58-oligoA
`synthetase enhancer, and a mutated variant were a generous gift
`from Shoumo Bhattacharya27 and were subcloned into pTGL2
`promoter vector (Promega, Madison, WI). BON cells (5 3 105
`cells/well) were plated in 6-well tissue culture dishes, and
`transfections were carried out by calcium phosphate precipita-
`tion technique using a DNA transfection kit (5 Prime-3 Prime;
`Boulder, CO), exactly as described previously.28 Transfected
`cells were allowed to recover for 16 hours and then stimulated
`for 24 hours with the indicated IFN-a concentrations. After
`lysis of cells, the luciferase activity was determined using
`luciferin, ATP, and coenzyme A (Promega) exactly as reported
`previously.28
`
`RNA Preparation and RT-PCR
`To confirm the presence of IFN-a receptor messenger
`RNA (mRNA) transcripts in QGP1 and BON cells, IFN-a
`receptor mRNA was amplified by RT-PCR. Total RNA from
`NE tumor cells was prepared using RNAzol R reagent (WAK
`Chemie, Bad Soden, Germany) according to the manufacturer’s
`instructions. RNA was submitted to deoxyribonuclease diges-
`tion, and aliquots of 1 µg were used for reverse transcription
`
`using Moloney murine leukemia virus reverse transcriptase.
`The reaction was diluted 1:25 for use in the subsequent PCR
`reaction with sequence-specific primers to a 639 base pair
`(bp)-spanning region of the human IFN-a receptor complemen-
`tary DNA (cDNA) (58-AGC GAT GAG TCT GTC GGG;
`38-GGC GTG GAG CCA CTG AAC). Amplification condi-
`tions were exactly as previously described.29
`Results
`Human NE Tumor Cells Express Functional
`IFN-a Receptors
`As a first step to establish the human endocrine
`GEP tumor cell lines QGP1 and BON as a suitable in
`vitro model to investigate IFN-a actions, we determined
`expression of IFN-a receptors at the mRNA level by
`RT-PCR analysis. Using sequence-specific primers against
`the IFN-a receptor cDNA, amplificates of the predicted
`size (639 bp) were detected in both NE cell lines as well
`as in Capan1 exocrine pancreatic cancer cells, which were
`included as positive control29 (Figure 1).
`To confirm that the mRNA transcripts corresponded
`to functionally active receptors, ligand-initiated signal
`transduction was examined next. IFN-a receptors consist
`of 2 chains that associate with Jak1 and Tyk2 kinases.
`Upon IFN-a binding to the receptor, Jak1 and Tyk2
`kinases approach each other and become activated by
`phosphorylation on tyrosine residues. To investigate
`IFN-a–dependent phosphorylation of JAKs in NE tumor
`cell lines, Jak1 (Figure 2, upper panel) and Tyk2 (Figure
`2, lower panel) were immunoprecipitated from whole-
`cell lysates and subsequently analyzed for tyrosine pho-
`phorylation by immunoblotting. After IFN-a stimula-
`tion, Jak1 immunoprecipitates from QGP 1 cells readily
`increased their complement of phosphotyrosine, starting
`as early as 1 minute and reaching a maximum at 15
`minutes. In contrast to the results obtained in QGP1
`
`Figure 1. NE tumor cells express IFN-a receptor mRNA. Total RNA was
`extracted from QGP1, BON, and Capan1 cell
`lines, reverse-tran-
`scribed, and amplified by PCR using primers directed against the type I
`IFN receptor a chain. For size determination of the obtained PCR
`fragment, a 100-bp DNA standard was used (lane 1). Alternating
`sample lanes represent reactions with (1) or without (2) addition of
`reverse transcriptase.
`
`Roxane Labs., Inc.
`Exhibit 1039
`Page 004
`
`

`
`April 2000
`
`IFN–a IN NEUROENDOCRINE TUMOR CELLS 739
`
`Figure 2. IFN-a treatment initiates activation of receptor-associated kinases in human NE tumor cells. Immunoblots of Jak1 and Tyk2
`immunoprecipitates analyzing IFN-a–induced changes in the phosphotyrosine content of IFN-a receptor–associated kinases in (A) QGP1 and (B)
`BON cells. Cells were incubated with 1000 IU/mL IFN-a for the indicated periods. Blots were subsequently stripped and reprobed with antibodies
`against Jak1 or Tyk2 to confirm that equal amounts of immunocomplexes were evaluated. Because Jak1 phosphotyrosine signals were notably
`weaker in BON cells than in QGP1 cells, the autoradiography had to be exposed for 30 minutes compared with 1–2-minute exposures required in
`QGP1 cells.
`
`cells, little increase in Jak1 tyrosine phosphorylation was
`observed in IFN-a–treated BON cells (Figure 2B),
`although weak tyrosine phosphorylation was consistently
`observed under control conditions. However, both QGP1
`and BON cells demonstrated a distinct, time-dependent
`increase in Tyk2 tyrosine phosphorylation, indicative of
`Tyk2 activation at 1, 5, and 15 minutes after IFN-a
`stimulation. Subsequent immunoblotting of the stripped
`phosphotyrosine blots with antibodies against Jak1 (Fig-
`ure 2, upper panel, second row) and Tyk2 (Figure 2, lower
`panel, second row) confirmed that equal amounts of
`immunocomplexes had been analyzed in IFN-a–treated
`cells and control cultures.
`In response to ligand-dependent IFN-a receptor activa-
`tion, latent cytoplasmic Stat1 and Stat2 transcription
`factors are recruited to specific binding sites on the
`cytoplasmic portion of the receptor, where Jak kinases
`then activate these Stat proteins via phosphorylation on
`tyrosine residues. Tyrosine phosphorylation of Stat pro-
`teins was therefore examined to confirm IFN-a–induced
`
`activation of downstream effector molecules. In QGP1
`(Figure 3A) and BON cells (Figure 3B), a pronounced,
`time-dependent increase in Stat 1 tyrosine phosphoryla-
`tion (Figure 3, upper panel) was evident. Enhanced
`phosphorylation was detected on both the 91- and
`84-kilodalton splice variants, with stronger signals for
`the 91-kilodalton form most likely reflecting its higher
`abundance in human NE cells (Figure 3, upper panel,
`second row). Similarly, phosphorylation of Stat2 was
`enhanced (Figure 3, lower panel), although the increase
`was moderate compared with the changes observed for
`Stat1. Comparison of the time courses of Stat activation
`in both cell lines revealed slightly different kinetics.
`Phosphorylation of Stat proteins in QGP1 was transient,
`with maximal effects occurring at 15 minutes of IFN-a
`stimulation for Stat1 and at 5 minutes for Stat2. In BON
`cells, phosphorylation of Stat1 and Stat2 was first evident
`at 5 minutes and thereafter persisted to 60 minutes with
`Stat1 phosphotyrosine levels showing a continuous in-
`crease over this period of time.
`
`Figure 3. IFN-a time-dependently induces tyrosine phosphorylation of Stat1 and Stat2 in NE tumor cells. Both tyrosine phosphorylated (upper
`row) and total (lowerrow). Stat complement of Stat1 (upperpanel) and Stat2 immunoprecipitates (lowerpanel) from IFN-a–treated cells were
`evaluated by immunoblotting. Cells were treated with 1000 IU/mL IFN-a for the indicated intervals. p91 and p84 indicate Stat 1 a- and b-splice
`variants, respectively, based on their molecular weight. Data are from representative experiments in (A) QGP1 and (B) BON cells.
`
`Roxane Labs., Inc.
`Exhibit 1039
`Page 005
`
`

`
`740 DETJEN ET AL.
`
`GASTROENTEROLOGY Vol. 118, No. 4
`
`After activation of latent Stat transcription factors by
`phosphorylation, Stat1 and Stat2 form heterodimers that
`translocate into the nucleus, where they initiate transcrip-
`tion from ISREs. To directly determine IFN-a–depen-
`dent transactivation of ISREs in NE cells, reporter gene
`studies were performed. In transient transfection experi-
`ments using BON cells, IFN-a dose-dependently stimu-
`lated transcription from an ISRE-luciferase reporter
`construct, but not from a mutant ISRE (Figure 4A),
`indicating that IFN-a–induced Stat signaling results in
`specific transactivation of IFN-regulated genes in this
`cell line. Because we repeatedly failed to achieve accept-
`able expression levels of the reporter construct in trans-
`fected QGP1 cells, nuclear translocation was determined
`as a substitute for reporter gene studies. IFN-a stimula-
`tion led to a time-dependent increase of Stat1 and Stat2
`proteins in nuclear extracts of QGP1 cells (Figure 4B),
`documenting the translocation of activated Stat proteins
`in response to IFN-a.
`IFN-a Inhibits Growth of Human NE
`Tumor Cells
`Having established an intact IFN-a–signaling
`machinery in both cell lines, we next addressed the
`question of whether IFN-a exhibits a direct antiprolifera-
`tive action on QGP1 and BON cells. Growth curves in
`the presence or absence of IFN-a were obtained over a
`period of 6 days (Figure 5A). IFN-a treatment (1000
`IU/mL) resulted in a significant inhibition of prolifera-
`tion in QGP1 (Figure 5A) and in BON cells (Figure 5B).
`The growth-inhibitory effects of IFN-a were concentra-
`tion dependent (Figure 5C and D) with half maximal
`effects on proliferation occurring at less than 100 IU/mL
`in QGP1 and at approximately 500 IU/mL in BON cells,
`which is within the therapeutically achievable range of
`plasma levels in humans.
`
`Growth of malignant tumors in vivo is influenced by
`unique properties of transformed cells, which are more
`closely reflected by anchorage-independent growth assays
`than by substrate-dependent proliferation assays. To
`establish that IFN-a is capable of directly regulating
`anchorage-independent growth of NE tumor cells, colony
`formation of QGP1 and BON cells in agar suspension
`was evaluated. IFN-a treatment profoundly inhibited
`substrate-independent growth of QGP1 cells in a dose-
`dependent manner (Figure 6A) with a maximal decrease
`in colony number to 15% 6 5% of control achieved with
`1000 IU/mL IFN-a. Similarly, substrate-independent
`growth of BON cells was impaired (Figure 6B), although
`the maximal reduction to 77% of control was only
`moderate and did not reach statistical significance. Thus,
`both substrate-dependent and substrate-independent
`growth of NE tumor cells were directly inhibited by
`IFN-a.
`IFN-a–Treated NE Tumor Cells Accumulate
`in the S Phase of the Cell Cycle
`Depending on the cell model investigated, various
`mechanisms responsible for the growth-inhibitory effects
`of IFN-a have been suggested, most of which involve
`modulation of Rb. To test whether Rb might also be
`involved in IFN-a–mediated growth inhibition of NE
`tumor cells,
`immunoblotting experiments were per-
`formed to analyze expression and phosphorylation status
`of Rb in QGP1 and BON cells (Figure 7). These blots
`revealed a striking difference in the Rb content of the 2
`cell lines. Bands representing the hypophosphorylated
`and hyperphosphorylated forms of Rb were easily de-
`tected both in BON and exocrine Capan1 cells, but no
`signal could be visualized in QGP1 cells when equivalent
`amounts of protein were examined. Control blots using
`an antibody against p16ink4a showed a complementary
`
`Figure 4. IFN-a stimulation results in transactivation of an ISRE-driven reporter construct. (A) Dose-dependent increase of relative luciferase
`activity (RLU) in IFN-a–treated BON cells transiently transfected with pGL2-ISRE-luc, but not mutant pGL2-DISRE-luc. Data represent means 6 SEM
`from at least 3 separate experiments conducted in triplicate. (B) Immunoblot analysis of Stat1 and Stat2 in nuclear extracts of QGP1 cells,
`confirming the nuclear translocation of Stat transcription factors in cells stimulated with 1000 IU/mL IFN-a for the indicated times.
`
`Roxane Labs., Inc.
`Exhibit 1039
`Page 006
`
`

`
`April 2000
`
`IFN–a IN NEUROENDOCRINE TUMOR CELLS 741
`
`Figure 5. IFN-a inhibits growth of NE tumor cells in a time- and dose-dependent manner. Subconfluent cells were treated with (A and B) 1000
`IU/mL IFN-a (d) or vehicle (s) for the indicated time periods or with (Cand D) 100 (h), 500 (j), and 1000 (D) IU/mL for the indicated times.
`Viable cells were then manually counted in a hemacytometer. Data represent means 6 SEM from at least 3–4 separate experiments, each
`conducted in triplicate. *P# 0.05 compared with vehicle-treated controls.
`
`expression pattern, as previously described for a variety of
`cells.30 Neither the expression level of p16ink4a nor the
`relative abundance of the hypophosphorylated form of Rb
`were up-regulated by IFN-a treatment, whereas overall
`pRb content was even moderately reduced at 48 hours
`(Figure 7), indicating that the growth inhibition ob-
`served in NE tumor cells did not require activation of Rb.
`To identify potential mechanisms responsible for the
`growth-regulatory effects of IFN-a in NE cells, IFN-a–
`induced cell cycle redistribution was determined by flow
`
`cytometry (Figure 8). In these experiments on unsynchro-
`nized cells, IFN-a treatment increased the fraction of
`cells in the S phase of the cell cycle, whereas the
`proportion of cells in the G1 phase decreased (Figure 8A).
`Both QGP1 and BON cell lines displayed a comparable
`redistribution pattern (Figure 8B), again supporting the
`notion that IFN-a elicits Rb-independent cell cycle–
`specific effects in NE cells. Of note, no population with
`
`Figure 6. IFN-a inhibits anchorage-independent growth of NE tumor
`cells. A single cell agar supension of (A) QGP1 or (B) BON cells was
`incubated with the indicated doses of IFN-a for 10 days, and colony
`formation was evaluated. Data represent means 6 SEM from at least
`3 separate experiments, each conducted in triplicate. *P# 0.05 and
`**P# 0.01 compared with vehicle-treated controls.
`
`Figure 7. IFN-a treatment does not affect Rb phosphorylation in NE
`tumor cells. (A) Immunoblot analysis of Rb expression and phosphory-
`lation status in control and IFN-a–stimulated NE tumor cells as well as
`Capan1 exocrine pancreatic cancer cells. In each lane, 15 µg of whole
`cell lysates was separated by 7.5% SDS-PAGE. (B) Aliquots of the
`same lysates immunoblotted for expression of the CKI p16 ink4a.
`
`Roxane Labs., Inc.
`Exhibit 1039
`Page 007
`
`

`
`742 DETJEN ET AL.
`
`GASTROENTEROLOGY Vol. 118, No. 4
`
`Figure 8. IFN-a–treated NE tumor cells accumulate in the S phase of
`the cell cycle. (A) Representative fluorescence-activated cell sorter
`analysis showing changes in cell cycle distribution of QGP1 cells
`treated with 1000 IU/mL IFN-a for 48 hours. (B) Summary of
`IFN-a–induced cell cycle redistribution in QGP1 and BON cells. Data
`are expressed as mean 6 SEM of the percentage of cells in the S
`phase (closedsymbols) and the G1 phase (opensymbols) compared
`with untreated control cultures, determined in 3 independent experi-
`ments. *P# 0.05 and **P# 0.01 compared with control.
`
`subdiploid DNA content was observed in these experi-
`ments, indicating that no relevant induction of apoptotic
`cell death had occurred. Analysis of the kinetics of
`IFN-a–induced cell cycle effects revealed first S-pha

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket